RESEARCH
Peer reviewed scientific video journal
Video encyclopedia of advanced research methods
Visualizing science through experiment videos
EDUCATION
Video textbooks for undergraduate courses
Visual demonstrations of key scientific experiments
BUSINESS
Video textbooks for business education
OTHERS
Interactive video based quizzes for formative assessments
Products
RESEARCH
JoVE Journal
Peer reviewed scientific video journal
JoVE Encyclopedia of Experiments
Video encyclopedia of advanced research methods
EDUCATION
JoVE Core
Video textbooks for undergraduates
JoVE Science Education
Visual demonstrations of key scientific experiments
JoVE Lab Manual
Videos of experiments for undergraduate lab courses
BUSINESS
JoVE Business
Video textbooks for business education
Solutions
Language
English
Menu
Menu
Menu
Menu
A subscription to JoVE is required to view this content. Sign in or start your free trial.
Research Article
Bingwei Wang1,2, Chunxiang Zhang1,2,3,4,5
1Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education,Southwest Medical University, 2Nucleic Acid Medicine, Key Laboratory of Luzhou,Southwest Medical University, 3Department of Cardiology, The Affiliated Hospital of Southwest Medical University,Southwest Medical University, 4Key Laboratory of Medical Electrophysiology, Ministry of Education,Southwest Medical University, 5Institute of Cardiovascular Research,Southwest Medical University
Erratum Notice
Important: There has been an erratum issued for this article. View Erratum Notice
Retraction Notice
The article Assisted Selection of Biomarkers by Linear Discriminant Analysis Effect Size (LEfSe) in Microbiome Data (10.3791/61715) has been retracted by the journal upon the authors' request due to a conflict regarding the data and methodology. View Retraction Notice
This protocol describes a step-by-step method for generating functional human cardiac organoids from human embryonic stem cells (hESCs). It includes cell thawing, differentiation, and maturation steps, providing a robust platform for cardiac development and disease modeling.
Human embryonic stem cell (hESC)-derived cardiac organoids are multicellular three-dimensional (3D) structures that recapitulate key aspects of early human heart development and function. These self-organizing organoids exhibit spontaneous contractility, cardiomyocyte marker expression, and tissue-like architecture reminiscent of native myocardium. Here, we present a robust and reproducible protocol to generate cardiac organoids from the H9 hESC line via stepwise lineage differentiation. Mesoderm induction is initiated by treating spheroids for 36-40 h with Activin A (50 ng/mL), bone morphogenetic protein 4 (BMP4, 10 ng/mL), fibroblast growth factor 2 (FGF2, 30 ng/mL), laduviglusib (CHIR99021, 3 µM), and a phosphatidylinositol 3-kinase (PI3K) inhibitor (LY294002, 5 µM). Cardiac lineage specification is subsequently directed by daily exposure for four days to BMP4 (10 ng/mL), FGF2 (10 ng/mL), a Wnt pathway inhibitor (XAV-939, 5 µM), and retinoic acid (0.5 µM). Cardiomyocyte differentiation and maturation are further promoted from Day 5.5 onward using BMP4 (10 ng/mL), FGF2 (10 ng/mL), and insulin (10 µg/mL). Functional validation is achieved through time-lapse imaging and immunofluorescence analysis, confirming the generation of contractile cardiomyocytes marked by cardiac troponin T (cTnT) expression. Additionally, 3D immunostaining reveals the presence of α-SMA and CDH5, indicating the emergence of smooth muscle and endothelial-like cell populations. These cardiac organoids consistently demonstrate rhythmic contractions; however, direct electrophysiological validation of electromechanical coupling was not performed. Known limitations include the inability to passage organoids and potential central necrosis during extended cultures. In summary, this model provides a scalable and physiologically relevant platform for studying human cardiogenesis, drug responses, and congenital heart diseases.
Cardiovascular disease remains the leading cause of morbidity and mortality worldwide, underscoring the urgent need for reliable in vitro models that recapitulate human cardiac development and function1,2. Traditional two-dimensional (2D) monolayer cultures of cardiomyocytes, while valuable, fail to reproduce the spatial complexity, multicellular interactions, and electromechanical integration of the native human heart3,4. 3D cardiac organoids derived from pluripotent stem cells have emerged as powerful platforms that address these limitations, providing self-organizing, multicellular structures that mimic early cardiac morphogenesis and functional maturation in vitro5.
hESCs, such as the H9 line, possess the intrinsic ability to differentiate into all somatic cell types, including cardiomyocytes, endothelial cells, and cardiac fibroblasts6. Under controlled culture conditions, hESCs can be guided through the sequential stages of mesoderm induction, cardiac mesoderm specification, and cardiomyocyte differentiation5,7. This stepwise process can be regulated using specific signaling molecules and inhibitors, including Activin A, BMP4, FGF2, PI3K inhibitors, CHIR99021, Wnt inhibitors, and retinoic acid5,8. By closely mimicking the in vivo signaling environment of embryonic cardiac development, these cues promote efficient and reproducible cardiogenesis in vitro5,7.
Unlike many organoid systems that require embedding in extracellular matrix (ECM) components such as Matrigel, cardiac organoids can self-assemble in low-adhesion conditions without matrix support5,9. Aggregation of differentiating hESCs in suspension yields spherical 3D structures that exhibit spontaneous beating within the first week of differentiation10. These organoids contain functionally relevant cardiac cell populations and demonstrate characteristic features, such as sarcomeric organization, calcium transients, and rhythmic contractions11,12, offering a physiologically relevant model of early human heart tissue.
The ability to generate hESC-derived cardiac organoids offers exciting opportunities for developmental biology, disease modeling, drug screening, and cardiotoxicity testing13,14. Moreover, integration with genome editing technologies and patient-specific induced pluripotent stem cells (iPSCs) enables personalized disease modeling and therapeutic exploration for inherited cardiac disorders.
In this article, we present a detailed, step-by-step protocol for the generation of cardiac organoids from human embryonic stem cells, highlighting key stages of differentiation, culture conditions, and validation techniques. This platform provides a robust, scalable system for recapitulating human cardiac development and establishing a foundation for future translational research.
All procedures involving human embryonic stem cells (H9) were conducted in accordance with institutional guidelines and approved by Biomedical Ethics Committee of Southwest Medical University, approval 20241023-031. All tissue culture work detailed below should be done in a Class II laminar flow hood. Always ensure that media and reagents are at room temperature, equilibrated naturally before use. Do not use a water bath to warm media.
1. Thawing and routine culture of HESCs (H9)
2. Differentiation of HESCs (H9) into cardiac lineage
3. Preparation for cryopreservation
4. Cardiac organoid recovery and thawing
The successful differentiation of H9 hESCs into cardiac organoids begins with the robust revival and maintenance of pluripotent stem cells. Upon thawing, hESCs demonstrate high viability and adherence when plated on vitronectin-coated plates in E8 medium supplemented with a ROCK inhibitor. Within 24-48 h, compact colonies with defined edges and high nucleus-to-cytoplasm ratios can be observed, indicating recovery of typical undifferentiated morphology (Figure 1A). After thawing, hESCs rapidly enter a proliferative phase, demonstrating robust colony expansion and re-establishment of pluripotent characteristics (Figure 1A).
The overall workflow for generating cardiac organoids from H9 hESCs is illustrated in Figure 1B, detailing each stage from stem cell maintenance to 3D organoid formation and maturation. The successful differentiation of hESCs into cardiac organoids is evident through a series of distinct morphological and functional transitions. Within the first 24 h following aggregation in ultra-low attachment plates, compact and uniform spheroids form. These spheroids exhibit a smooth, round morphology with high optical density, indicating high cell viability and homogeneous aggregation (Figure 1C).
By Day 4-5 of mesodermal induction, the spheroids begin to display a flattened and slightly expanded appearance, reflecting cellular differentiation and structural remodeling (Figure 1C). From Day 7 onward, spontaneous contractions are typically observed in over 70% of aggregates, indicating the onset of cardiomyocyte functionality (Figure 1C and Video 1).
By Day 12+, mature cardiac organoids demonstrate synchronous and rhythmic contractions. Immunofluorescence staining confirms the expression of the cardiac-specific marker cardiac troponin T (cTnT), which is distributed throughout the cardiac organoids (Figure 2A), indicating the presence of functionally relevant cardiomyocyte populations. Three-dimensional immunofluorescence staining further demonstrates the presence of vascular and smooth muscle lineage components within the cardiac organoids. CDH5 (VE-cadherin), a marker of endothelial cells, is broadly detected across the organoid surface and within interior regions, suggesting endothelial network formation. Concurrently, α-smooth muscle actin (αSMA) expression is observed in a subset of cells (Figure 2B). The co-expression of αSMA and CDH5 in spatially distinct domains highlights the multicellular complexity and tissue-like architecture of the cardiac organoids. In addition, quantitative analysis reveals that approximately 63% of cells within the cardiac organoids are positive for cTnT, 17% are positive for αSMA, and 9% express CDH5. These proportions are consistently observed across three independent experimental batches, demonstrating the robustness and reproducibility of the differentiation protocol.
Following isoproterenol (ISO)-induced injury, cardiac organoids exhibited altered expression of several cardiac markers. Quantitative PCR analysis showed a notably higher expression of MYH7 compared to MYH6 (Figure 3), consistent with a ventricular-like transcriptional profile. Additionally, elevated levels of ANP and BNP mRNA suggested activation of cardiac stress pathways. These results suggest that the organoids respond physiologically to injury and contain functionally relevant cardiomyocyte subtypes.
Organoids subjected to drug treatments or electrical stimulation may display altered beating rates or arrhythmic patterns, providing a platform for pharmacological testing or disease modeling. Moreover, cryopreserved organoids retain beating capacity upon thawing, though a brief recovery period (24-48 h) is required before functional assessments can be made.

Figure 1: Establishment and morphological progression of cardiac organoids from hESCs (H9). (A) Representative phase-contrast images showing the recovery process of hESCs at day 1, day 2, and day 3 post-thawing.(B) Schematic workflow outlining the stepwise differentiation protocol used for the generation of cardiac organoids from hESCs.(C) Brightfield images showing the morphological characteristics of cardiac organoids at key differentiation stages. (D) Spontaneous beating frequency of cardiac organoids. Cardiac organoids derived from H9 hESCs were monitored under brightfield time-lapse microscopy. Beating frequency was quantified on Day 12 by counting rhythmic contractions per minute in individual organoids. Data are presented as beats per minute (bpm), with each dot representing a single organoid. Error bars indicate mean ± standard error of the mean (SEM). Scale bars = 100 µm (A,C). Abbreviation: hESCs = human embryonic stem cells. Please click here to view a larger version of this figure.

Figure 2: Immunofluorescence characterization of cardiac organoids. (A) Immunofluorescence staining for cTnT demonstrates the presence of cardiomyocytes within the cardiac organoids at day 12.(B) 3D immunofluorescence staining shows positive expression of aSMA and CDH5, indicating the emergence of smooth muscle and endothelial-like cell populations, respectively. Scale bars = 100 µm (A), 50 µm (B). Abbreviations: cTnT = cardiac troponin T; aSMA = smooth muscle actin alpha; CDH5 = cadherin-5. Please click here to view a larger version of this figure.

Figure 3. Expression of cardiac marker genes MYH6, MYH7, ANP, and BNP in cardiac organoids following ISO-induced injury. Quantitative PCR data are shown as mean ± SEM (n = 3 biological replicates). Statistical analysis was performed using one-way ANOVA followed by Dunnett's post hoc test for multiple comparisons. P < 0.05 was considered statistically significant (*P < 0.05, **P < 0.01, ***P < 0.001). Abbreviation: ISO = isoproterenol. Please click here to view a larger version of this figure.
Video 1: Spontaneous contractile activity of cardiac organoids. Please click here to download this File.
The successful generation of cardiac organoids from H9 hESCs relies on precise temporal and spatial regulation of key signaling pathways to mimic early cardiac development. This protocol recapitulates the critical stages of mesoderm induction, cardiac lineage specification, and self-organization into 3D contractile structures in a chemically defined system. The method provides a robust and reproducible platform for modeling human heart development, congenital heart disease, and cardiotoxicity screening.
One of the most critical steps in this protocol is ensuring the health and quality of the H9 hESCs prior to differentiation 5. Cells must be cultured to approximately 70% confluency and passaged at least once after thawing before differentiation is initiated15,16. Using freshly thawed hESCs without passaging often results in poor viability and inefficient cardiac induction. Additionally, during routine maintenance, overdigestion or excessive pipetting should be avoided to preserve colony integrity; optimal results are achieved when cells are maintained in small clusters of 2-10 cells. Avoid enzymatic passaging to maintain pluripotency and colony morphology. Use the stem cell dissociation reagent to preserve cell-cell contact and minimize stress, but avoid overdigestion or excessive pipetting that can result in a single-cell suspension.
To ensure reproducibility and scalability, several experimental constraints and quality control measures must be considered. In particular, cell viability should exceed 85% post-thaw, and differentiation should not be attempted directly after thawing. Cells should undergo at least one passage before induction. Uniform spheroid formation is essential; compact embryoid bodies (EBs) should form within 24 h of aggregation in ultra-low attachment plates. Spontaneous beating typically begins by Day 7.5, and failure to observe contractions in >70% of organoids may indicate protocol deviation or suboptimal cell quality. These metrics help standardize outcomes across laboratories and facilitate broader adoption of this cardiac organoid model.
Another essential factor is the precise composition and timing of media components. MD-M, enriched with Activin A, BMP4, FGF2, CHIR99021, and a PI3K inhibitor, initiates efficient induction of mesodermal fate7. The subsequent stages use Wnt inhibitors and retinoic acid to promote cardiac specification and maturation. The use of a chemically defined medium throughout the protocol minimizes batch variability and supports consistent cardiac differentiation outcomes5.
Unlike other organoid systems, cardiac organoids do not support passaging or serial expansion5,17, mainly due to the limited proliferative capacity of cardiomyocytes, even at immature stages. Attempts to passage or mechanically dissociate cardiac organoids often result in loss of contractile function and structural integrity. Therefore, it is recommended that aggregates be formed at optimized cell densities and maintained without physical disturbance during the beating stages.
Compared to existing cardiac organoid protocols9, this method offers several notable advantages. First, it utilizes a chemically defined culture system with standardized and commercially available components, thereby minimizing batch-to-batch and inter-experimental variability and enhancing reproducibility. Second, the protocol supports cryopreservation of cardiac organoids with robust post-thaw functional recovery, enabling long-term storage and longitudinal analysis. Collectively, these features contribute to the reliability and consistency of cardiac organoid generation, making this protocol particularly suitable for scalable applications and comparative studies.
In addition, it avoids the use of undefined factors such as serum or feeder layers, enabling more standardized and scalable cardiac differentiation. It faithfully generates 3D structures that spontaneously contract and express cardiac markers, providing a physiologically relevant system for studying human-specific cardiac development. The ability to cryopreserve and recover beating cardiac organoids further enhances the versatility of this system for longitudinal studies or high-throughput applications.
In this study, cardiac organoids were cryopreserved using a specialized freezing medium and a standard slow-freezing protocol. Upon thawing, the organoids were maintained in maturation medium for continued culture. Survival rate was assessed 48 h post thaw by calculating the proportion of organoids exhibiting both intact morphology and spontaneous beating, as observed under a light microscope. Although we did not directly compare the proportions of cardiomyocytes, smooth muscle cells, and endothelial cells before and after freezing, we observed that the vast majority of cardiac organoids gradually regained spontaneous contractile activity within 3-4 days post thaw. Furthermore, they maintained visible beating behavior throughout the approximately 14 day observation period. However, compared to the pre-freezing state, thawed organoids generally exhibited a reduction in both beating frequency and contractile strength, indicating a trend toward diminished functional activity. These findings suggest that, although the cryopreserved organoids retain basic structural integrity and functionality after recovery, their physiological activity may be partially compromised. This highlights the need for further optimization of cryopreservation strategies to better preserve functional performance.
Despite its robustness, this protocol is not without limitations. Some degree of variability may arise due to differences in cell line behavior or environmental factors such as CO2 levels and humidity. In addition, long-term culture beyond 12 days may lead to central necrosis or fibrosis-like features due to limited nutrient and oxygen diffusion, especially in the absence of vascularization. Co-culture with endothelial cells or application of microfluidic perfusion systems may improve these limitations in future adaptations. Further, these cardiac organoids consistently exhibited rhythmic and spontaneous contractile activity, suggesting the presence of functional cardiomyocyte populations; however, direct electrophysiological assessments to confirm definitive electromechanical coupling were not conducted in the current study. Moreover, this study was primarily designed and optimized using the H9 hESC line, which is a widely accepted standard model in the field of cardiac differentiation and has been extensively validated for its reliable differentiation efficiency and stability. Therefore, at this stage of the research, we did not conduct validation experiments using additional cell lines. In future studies, we plan to extend this work to include other hESC or induced pluripotent stem cell (iPSC) lines to further enhance the generalizability and applicability of our findings.
While we did not perform systematic subtype characterization of atrial versus ventricular cardiomyocytes, our ISO-injury experiments revealed a significantly higher MYH7-to-MYH6 expression ratio, consistent with a ventricular-like profile, alongside increased ANP and BNP expression as markers of cardiac stress. These results indicate that the cardiac organoids not only contain functional cardiomyocyte populations but also exhibit physiologically relevant stress responses. Nonetheless, more comprehensive analyses, including immunostaining and electrophysiological profiling, will be required to fully delineate subtype identities and functional heterogeneity in future studies.
Although cryopreserved organoids demonstrated survival and spontaneous beating upon recovery, a systematic comparison of cellular composition before and after freezing was not performed in this study. This represents a limitation, as potential shifts in the relative proportions of cardiomyocytes, smooth muscle cells, and endothelial-like populations may have occurred but were not quantified. Future studies incorporating flow cytometry, immunostaining, or single-cell RNA sequencing will be necessary to rigorously evaluate the impact of cryopreservation on cell-type composition and functional integrity.
In conclusion, this method provides a simple, reproducible, and efficient strategy for generating human cardiac organoids from pluripotent stem cells. It is a valuable tool for developmental biology, disease modeling, and cardiotoxicity studies. Future improvements, such as the incorporation of vascular networks or innervation, may enhance the physiological relevance of the model even further.
The authors have no conflicts of interest to declare.
We thank Junfeng Ji (School of Basic Medical Science, Zhejiang University, China) and Tao Luo (School of Basic Medical Science, Zhejiang University, China) for kindly providing hESCs (H9). This work was supported by grants from the National Natural Science Foundation of China (Grant No. U23A20398 to C.Z.), Sichuan Science and Technology Program (Grant No. 2022YFS0578 and 2022YFS0614 to C.Z.), Research Start-up Foundation of Southwest Medical University (Grant No. 00040155 to C.Z.), Research Start-up Foundation of Southwest Medical University (Grant No. 00170071 to B.W.), The Science and Technology Strategic Cooperation Programs of Luzhou Municipal People's Government and Southwest Medical University (Grant No. 2024LZXNYDJ088 to B.W.), the Science and Technology Strategic Cooperation Project of Southwest Medical University (Grant No. 2024PZXNYD01 to BW), and the Foundation of Southwest Medical University (Grant No. 2024ZKZ014 to B.W.).
| 0.02% EDTA | Beyotime | C0198 | |
| 6 well plate | CORNING | 3516 | |
| 7.5% bovine serum albumin | ACMEC | AC11954 | |
| 96-well ultra-low attachment plate | CORNING | 7007 | |
| Alexa Fluor 488 Donkey anti-Mouse IgG | Yeasen | 34106ES60 | |
| Alexa Fluor 555 Goat anti-rabbit | Invitrogen | A-21428 | |
| CHIR-99021 monohydrochloride | Yeasen | 52965ES10 | |
| DMSO | Solarbio | D8370 | |
| Dulbecco Phosphate-Buffered Saline (D-PBS) | Yeasen | 60152ES76 | |
| FBS | Gibco | 10099141C | |
| FGF2 Protein, Human, Recombinant | TargetMol | TMPY-00749-50 μg | |
| Ham's F-12 Nutrient Mix | life technologies | 11765054 | |
| hPSC-CDM (Essential 8 medium) | Cauliscell | 400105 | |
| Human BMP-4 Recombinant Protein | Gibco | PHC9534 | |
| IMDM, GlutaMAX Supplement | life technologies | 31980030 | |
| Insulin-Transferrin-Selenium (ITS -G) | Gibco | 41400045 | |
| LY294002 | Selleck | s1105-50mg | |
| Monothioglycerol | Sigma-Aldrich | M6145 | |
| Mouse anti-αSMA | Abmart | MN50104 | |
| ProLong Diamond Antifade Mountant with DAPI | Invitrogen | P36966 | |
| Rabbit anti-CDH5 | Abmart | TA6265 | |
| Rabbit anti-cTNT | Invitrogen | 701620 | |
| Recombinant Human BMP-4 Protein | Yeasen | 92053ES20 | |
| Recombinant Human Insulin | Yeasen | 40112ES25 | |
| Recombinant Human/Mouse/Rat Activin A Protein | Yeasen | 91702ES10 | |
| ReLeSR | Stemcell | 100-0483 | |
| Retinoic Acid | Sigma-Aldrich | R2625 | |
| Vitronectin | Gibco | A14700 | |
| XAV-939 | Selleck | S1180 | |
| Y-27632 2HCl (ROCK inhibitor) | Selleck | S1049 |